Na-Cl co-transporter (NCC) is a 125-kDa 12-transmembrane Na+-dependent cation-chloride co-transporter that is primarily expressed in kidney distal convoluted tubules to resorb the filtered load of NaCl in the kidney. In mice, deficiency of NCC causes Gitelman syndrome, an inherited hypokalemic alkalosis with significant hypocalciuria and hypomagnesemia, which are secondary to the deficit in NaCl resorption. Interleukin 18 (IL18) is an inflammatory cytokine that promotes inflammatory cell cytokine/chemokine (e.g., IFN-?, IL6, MCP-1) production. In atherosclerotic lesions, increased levels of IL18 and its cognate receptor IL18r are detected in macrophages, smooth muscle cells (SMCs), and endothelial cells (ECs). Serums IL18 levels are elevated in patients with CAD, correlate with CAD severity score, are associated with cardiovascular risk factors, and predict future CAD events. In experimental atherosclerosis, overexpression or intraperitoneal administration of IL18 increases atherosclerotic lesion formation and enhances vulnerable plaque phenotypes via an IFN-?- dependent pathway, while inactivation or genetic deficiency of IL18 slows lesion progression. Our preliminary studies show that IL18 binds to NCC and mediates downstream protein tyrosine phosphorylation in mouse heart ECs. Stimulation of macrophages, T cells, and aortic SMCs and ECs with inflammatory cytokines (e.g., IL18, TNF-?, IL1) induces NCC expression. In both humans and mice, normal aortas express negligible NCC and IL18r, but SMCs, ECs, and macrophages in atherosclerotic lesions express high amounts of both NCC and IL18r. In atherosclerosis-prone apolipoprotein E-deficient (Apoe-/-) mice, the absence of either IL18r or NCC does not slow atherosclerosis development significantly. But combined deficiency of IL18r and NCC significantly reduces aortic arch macrophage content and lesion areas, and thoracic-abdominal aorta lipid deposition, along with reduced serum cytokines IFN-? and IL6. From cultured macrophages, the combined absence of NCC and IL18r impairs IL18-mediated cell signaling, and significantly reduces IL18 cell-surface binding and consequent inflammatory cytokine and chemokine production. These observations lead to our central hypothesis that NCC and IL18r are alternative IL18-binding molecules on inflammatory cells and cardiovascular cells, and that they mediate IL18 activities cooperatively in atherogenesis and possibly in other inflammatory diseases. We propose three aims: 1). to examine whether inhibition, activation, or genetic deficiency of NCC affects atherogenesis; whether NCC activities on inflammatory cells (e.g. macrophages) are essential to atherogenesis; and age-dependent; 2). To examine whether NCC acts like other kinase receptor, NCC activities in salt uptake and IL18 signaling interplay with each other, IL18 has different affinity for NCC and IL18r, and NCC is specific for IL18; and 3). To examine whether NCC mediates IL18 activities in T cells and vascular SMCs and ECs.

Public Health Relevance

Na-Cl co-transporter (NCC) is a 12-transmembrane cation-chloride co-transporter, expressed in the kidney distal convoluted tubules. We discovered that this ion channel co-transporter mediates IL18 signaling and cytokine production in ECs, macrophages, and NCC-transfected COS-7 cells. Induced NCC expression in macrophages, T cells, and vascular SMCs and ECs after inflammatory cytokine stimulation, increased NCC expression in atherosclerotic lesions, and impaired atherosclerosis in IL18-deficient Apoe-/- mice, but not in cognate IL18 receptor IL18r-deficient Apoe-/- mice, leading to our central hypothesis that this cell-surface ion channel protein is a novel alternative IL18-binding protein and cooperates with IL18r in mediating IL18 inflammation during atherogenesis, and possibly in other IL18-associated inflammatory diseases.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
1R01HL123568-01A1
Application #
8882740
Study Section
Special Emphasis Panel (ZRG1-CB-G (55))
Program Officer
Hasan, Ahmed AK
Project Start
2015-08-21
Project End
2019-05-31
Budget Start
2015-08-21
Budget End
2016-05-31
Support Year
1
Fiscal Year
2015
Total Cost
$420,408
Indirect Cost
$170,408
Name
Brigham and Women's Hospital
Department
Type
DUNS #
030811269
City
Boston
State
MA
Country
United States
Zip Code
02115
Wang, Yunzhe; Liu, Cong-Lin; Lindholt, Jes S et al. (2018) Plasma Cystatin B Association With Abdominal Aortic Aneurysms and Need for Later Surgical Repair: A Sub-study of the VIVA Trial. Eur J Vasc Endovasc Surg 56:826-832
Tang, Ting-Ting; Li, Yuan-Yuan; Li, Jing-Jing et al. (2018) Liver-heart crosstalk controls IL-22 activity in cardiac protection after myocardial infarction. Theranostics 8:4552-4562
Yan, Xiang; Wu, Chun; Chen, Tao et al. (2017) Cathepsin S inhibition changes regulatory T-cell activity in regulating bladder cancer and immune cell proliferation and apoptosis. Mol Immunol 82:66-74
Zhou, Yi; Chen, Huimei; Liu, Li et al. (2017) Cathepsin K Deficiency Ameliorates Systemic Lupus Erythematosus-like Manifestations in Faslpr Mice. J Immunol 198:1846-1854
Liu, Cong-Lin; Santos, Marcela M; Fernandes, Cleverson et al. (2017) Toll-like receptor 7 deficiency protects apolipoprotein E-deficient mice from diet-induced atherosclerosis. Sci Rep 7:847
Zhu, Zhaozhong; Liang, Liming; Zhang, Ruyang et al. (2017) Whole blood microRNA markers are associated with acute respiratory distress syndrome. Intensive Care Med Exp 5:38
Jin, Dong-Yi; Liu, Cong-Lin; Tang, Jun-Nan et al. (2017) Interleukin-18, matrix metalloproteinase-22 and -29 are independent risk factors of human coronary heart disease. J Zhejiang Univ Sci B 18:685-695
Zhou, Yi; Chen, Huimei; Liu, Li et al. (2017) CD74 Deficiency Mitigates Systemic Lupus Erythematosus-like Autoimmunity and Pathological Findings in Mice. J Immunol 198:2568-2577
Liu, Cong-Lin; Zhang, Jin-Ying; Shi, Guo-Ping (2016) Interaction between allergic asthma and atherosclerosis. Transl Res 174:5-22
Liu, Cong-Lin; Wang, Yi; Liao, Mengyang et al. (2016) Allergic lung inflammation promotes atherosclerosis in apolipoprotein E-deficient mice. Transl Res 171:1-16

Showing the most recent 10 out of 14 publications